Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Placenta ; 65: 7-14, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29908644

RESUMEN

INTRODUCTION: Several reports demonstrated that cadmium (Cd) had proinflammatory activities. The present study aimed to investigate whether Cd induces inflammatory cytokines in mouse placenta and human trophoblast cells. METHODS: Human JEG-3 cells were treated with different concentration of CdCl2 (0-50 µM) or CdCl2 (25 µM) for different times. The pregnant mice were administered with CdCl2 (3.0 mg/kg, i.p.) on GD15. RESULTS: TNF-α, IL-8 and IL-6 mRNAs were elevated in CdCl2-treated JEG-3 cells. Several inflammatory cytokines were up-regulated in Cd-treated placenta of mice. Moreover, keratinocyte chemokine (KC), a functional analogue of human IL-8, was increased in maternal serum and amniotic fluid from CdCl2-exposed mice. Additional experiment showed that gestational Cd exposure activated Akt signaling in mouse placenta. Co-culture with CdCl2 elevated pAkt level in JEG-3 cells in concentration- and time-dependent manners. LY294002, a specific inhibitor of PI3K, blocked CdCl2-evoked Akt phosphorylation in JEG-3 cells. Concomitantly, LY294002 inhibited CdCl2-induced IL-8 in JEG-3 cells. N-acetylcysteine (NAC), an antioxidant and a glutathione precursor, blocked CdCl2-evoked Akt phosphorylation in mouse placenta and human trophoblast cells. Additionally, NAC attenuated Cd-induced up-regulation of KC in amniotic fluid. DISCUSSION: Cd induces inflammatory cytokines partially through activating Akt signaling in mouse placenta and human trophoblast cells. NAC may be exploited for prevention of Cd-induced placental inflammation.


Asunto(s)
Cadmio/farmacología , Citocinas/genética , Mediadores de Inflamación/metabolismo , Placenta/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Trofoblastos/efectos de los fármacos , Animales , Células Cultivadas , Citocinas/metabolismo , Femenino , Humanos , Inflamación/genética , Inflamación/metabolismo , Masculino , Ratones , Placenta/metabolismo , Embarazo , Transducción de Señal/efectos de los fármacos , Trofoblastos/metabolismo , Regulación hacia Arriba/efectos de los fármacos
2.
PLoS One ; 13(1): e0191667, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29373603

RESUMEN

Cadmium (Cd) is a developmental toxicant that induces fetal growth restriction (FGR). Placental endoplasmic reticulum (ER) stress is associated with FGR. This study investigated the effects of N-acetylcysteine (NAC) on Cd-induced placental ER stress and FGR. Pregnant mice were intraperitoneally injected with CdCl2 daily from gestational day (GD)13 to GD17. As expected, Cd reduced fetal weight and crown-rump length. Cd decreased the internal space of blood vessels in the placental labyrinth layer and inhibited placental cell proliferation. Several genes of growth factors, such as Vegf-a, placental growth factor, Igf1 and Igf1r, and several genes of nutrient transport pumps, such as Glut1, Fatp1 and Snat2, were down-regulated in placenta of Cd-treated mice. Moreover, Cd evoked placental ER stress. Of interest, NAC alleviated Cd-induced FGR. Additional experiment showed that NAC inhibited Cd-induced impairment of placental development and placental ER stress. Therefore, NAC may be exploited for prevention of Cd-induced placental insufficiency and FGR.


Asunto(s)
Acetilcisteína/farmacología , Cadmio/toxicidad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Retardo del Crecimiento Fetal/prevención & control , Placenta/efectos de los fármacos , Animales , Regulación hacia Abajo/efectos de los fármacos , Femenino , Ratones , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa
3.
PLoS One ; 12(8): e0182584, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28783760

RESUMEN

Previous studies demonstrated that inflammatory microenvironment promoted prostate cancer progression. This study investigated whether total glucosides of paeony (TGP), the active constituents extracted from the root of Paeonia Lactiflora Pall, suppressed lipopolysaccharide (LPS)-stimulated proliferation, migration and invasion in androgen insensitive prostate cancer cells. PC-3 cells were incubated with LPS (2.0 µg/mL) in the absence or presence of TGP (312.5 µg /mL). As expected, cells at S phase and nuclear CyclinD1, the markers of cell proliferation, were increased in LPS-stimulated PC-3 cells. Migration activity, as determined by wound-healing assay and transwell migration assay, and invasion activity, as determined by transwell invasion assay, were elevated in LPS-stimulated PC-3 cells. Interestingly, TGP suppressed LPS-stimulated PC-3 cells proliferation. Moreover, TGP inhibited LPS-stimulated migration and invasion of PC-3 cells. Additional experiment showed that TGP inhibited activation of nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK)/p38 in LPS-stimulated PC-3 cells. Correspondingly, TGP attenuated upregulation of interleukin (IL)-6 and IL-8 in LPS-stimulated PC-3 cells. In addition, TGP inhibited nuclear translocation of signal transducer and activator of transcription 3 (STAT3) in LPS-stimulated PC-3 cells. These results suggest that TGP inhibits inflammation-associated STAT3 activation and proliferation, migration and invasion in androgen insensitive prostate cancer cells.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Glucósidos/farmacología , Lipopolisacáridos/farmacología , Paeonia/química , Neoplasias de la Próstata Resistentes a la Castración/patología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Masculino , FN-kappa B/metabolismo , Invasividad Neoplásica , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
Oncotarget ; 8(13): 22076-22085, 2017 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-28423553

RESUMEN

Vitamin D deficiency has been associated with increased risks of prostate cancer. Nevertheless, the mechanisms remain unclear. The aim of this study was to analyze the association among prostate cancer, vitamin D status and inflammation. Sixty patients with newly diagnosed prostate cancer and 120 age-matched controls were recruited for this study. Vitamin D status was evaluated and serum inflammatory molecules were measured. Serum 25-(OH)D was lower in patients with prostate cancer. Moreover, serum 25(OH)D was lower in patients with severe prostate cancer than patients with mild and moderate prostate cancer. By contrast, serum C-reactive protein (CRP) and interleukin (IL)-8, two inflammatory molecules, were elevated in patients with prostate cancer. Serum 25-(OH)D was negatively correlated with serum CRP and IL-8 in patients with prostate cancer. Additional analysis showed that the percentage of vitamin D receptor positive nucleus in the prostate was reduced in patients with prostate cancer. By contrast, the percentage of nuclear factor kappa B p65-positive nucleus was elevated in patients with prostate cancer. Our results provide evidence that there is an association among prostate cancer, vitamin D deficiency and inflammatory signaling. Inflammation may be an important mediator for prostate cancer progression in patients with low vitamin D status.


Asunto(s)
Biomarcadores de Tumor/sangre , Inflamación/sangre , Neoplasias de la Próstata/complicaciones , Deficiencia de Vitamina D/sangre , Vitamina D/sangre , Anciano , Proteína C-Reactiva/metabolismo , Estudios de Casos y Controles , Estudios de Seguimiento , Humanos , Inflamación/etiología , Masculino , Clasificación del Tumor , Pronóstico , Receptores de Calcitriol/sangre , Factores de Riesgo , Deficiencia de Vitamina D/etiología
5.
Am J Reprod Immunol ; 77(3)2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28045211

RESUMEN

PROBLEM: Increasing evidence demonstrates that inflammatory cytokines are involved in LPS-induced adverse pregnant outcomes including early embryo loss. Vitamin D3 (VitD3) has anti-inflammatory activity. We aimed to investigate the effects of vitamin D3 (VitD3) on LPS-induced early embryo loss in mice. METHOD OF STUDY: All pregnant mice except controls were intraperitoneally (ip) injected with LPS on GD7. In VitD3 alone and LPS+VitD3 groups, pregnant mice were pretreated with VitD3 by gavage daily from GD5 to GD7. RESULTS: LPS caused 62.5% pregnant mice with early embryo loss. Interestingly, the rate of abortion dropped to 14.3% when pregnant mice were pretreated with VitD3. Additional experiment showed that VitD3 significantly attenuated LPS-evoked elevation on TNF-α, IFN-γ, MIP-2, and nitrate plus nitrite in maternal serum. In addition, VitD3 alleviated LPS-induced COX-2 expression in the decidua and attenuated the elevation of PGF2α in maternal serum. Although VitD3 had no effect on IL-10 in maternal serum, it induced further elevation of serum IL-10 level in LPS-treated mice. Further analysis showed that VitD3 activated VDR signaling, simultaneously inhibited LPS-induced nuclear translocation of NF-κB p65 subunits in the decidua. CONCLUSIONS: VitD3 protects mice from LPS-induced early embryo loss at least partially through its anti-inflammatory effects.


Asunto(s)
Antiinflamatorios/inmunología , Colecalciferol/inmunología , Pérdida del Embrión/prevención & control , Inflamación/inmunología , Administración Oral , Animales , Ciclooxigenasa 2/metabolismo , Citocinas/sangre , Decidua/metabolismo , Dinoprost/sangre , Pérdida del Embrión/inmunología , Femenino , Humanos , Mediadores de Inflamación/sangre , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos ICR , FN-kappa B/metabolismo , Embarazo
6.
J Steroid Biochem Mol Biol ; 152: 133-41, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26013770

RESUMEN

Increasing evidence demonstrates that reactive oxygen species plays important roles in sepsis-induced acute kidney injury. This study investigated the effects of VitD3 pretreatment on renal oxidative stress in sepsis-induced acute kidney injury. Mice were intraperitoneally injected with lipopolysaccharide (LPS, 2.0mg/kg) to establish an animal model of sepsis-induced acute kidney injury. In VitD3+LPS group, mice were orally pretreated with three doses of VitD3 (25 µg/kg) at 1, 24 and 48 h before LPS injection. As expected, oral pretreatment with three daily recommended doses of VitD3 markedly elevated serum 25(OH)D concentration and efficiently activated renal VDR signaling. Interestingly, LPS-induced renal GSH depletion and lipid peroxidation were markedly alleviated in VitD3-pretreated mice. LPS-induced serum and renal nitric oxide (NO) production was obviously suppressed by VitD3 pretreatment. In addition, LPS-induced renal protein nitration, as determined by 3-nitrotyrosine residue, was obviously attenuated by VitD3 pretreatment. Further analysis showed that LPS-induced up-regulation of renal inducible nitric oxide synthase (inos) was repressed in VitD3-pretreated mice. LPS-induced up-regulation of renal p47phox and gp91phox, two NADPH oxidase subunits, were normalized by VitD3 pretreatment. In addition, LPS-induced down-regulation of renal superoxide dismutase (sod) 1 and sod2, two antioxidant enzyme genes, was reversed in VitD3-pretreated mice. Finally, LPS-induced tubular epithelial cell apoptosis, as determined by TUNEL, was alleviated by VitD3 pretreatment. Taken together, these results suggest that VitD3 pretreatment alleviates LPS-induced renal oxidative stress through regulating oxidant and antioxidant enzyme genes.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Colecalciferol/uso terapéutico , Riñón/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , 25-Hidroxivitamina D 2/sangre , Lesión Renal Aguda/inducido químicamente , Animales , Apoptosis/efectos de los fármacos , Glutatión/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Lipopolisacáridos , Masculino , Glicoproteínas de Membrana/biosíntesis , Ratones , NADPH Oxidasa 2 , NADPH Oxidasas/biosíntesis , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Receptores de Calcitriol/metabolismo , Superóxido Dismutasa/biosíntesis , Superóxido Dismutasa-1 , Tirosina/análogos & derivados , Tirosina/análisis
7.
Endocrinology ; 156(6): 2103-13, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25774554

RESUMEN

It is increasingly recognized that vitamin D deficiency is associated with increased risks of metabolic disorders among overweight children. A recent study showed that vitamin D deficiency exacerbated inflammation in nonalcoholic fatty liver disease through activating toll-like receptor 4 in a high-fat diet (HFD) rat model. The present study aimed to further investigate the effects of vitamin D deficiency on HFD-induced insulin resistance and hepatic lipid accumulation. Male ICR mice (35 d old) were randomly assigned into 4 groups as follows. In control diet and vitamin D deficiency diet (VDD) groups, mice were fed with purified diets. In HFD and VDD+HFD groups, mice were fed with HFD. In VDD and VDD+HFD groups, vitamin D in feed was depleted. Feeding mice with vitamin D deficiency diet did not induce obesity, insulin resistance, and hepatic lipid accumulation. By contrary, vitamin D deficiency markedly alleviated HFD-induced overweight, hyperinsulinemia, and hepatic lipid accumulation. Moreover, vitamin D deficiency significantly attenuated HFD-induced up-regulation of hepatic peroxisome proliferator-activated receptor γ, which promoted hepatic lipid uptake and lipid droplet formation, and its target gene cluster of differentiation 36. In addition, vitamin D deficiency up-regulated carnitine palmitoyltrans 2, the key enzyme for fatty acid ß-oxidation, and uncoupling protein 3, which separated oxidative phosphorylation from ATP production, in adipose tissue. These data suggest that vitamin D deficiency is not a direct risk factor for obesity, insulin resistance, and hepatic lipid accumulation. Vitamin D deficiency alleviates HFD-induced overweight, hyperinsulinemia, and hepatic lipid accumulation through promoting fatty acid ß-oxidation and elevating energy expenditure in adipose tissue.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Hiperinsulinismo/etiología , Hígado/efectos de los fármacos , Hígado/metabolismo , Deficiencia de Vitamina D/fisiopatología , Animales , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Energía/efectos de los fármacos , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos ICR , Obesidad/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Deficiencia de Vitamina D/metabolismo
8.
Toxicol Sci ; 145(1): 90-7, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25673501

RESUMEN

Several reports demonstrated that maternal lipopolysaccharide (LPS) exposure at middle gestational stage caused neural tube defects (NTDs). This study investigated the effects of supplementation with vitamin D3 (VitD3) during pregnancy on LPS-induced NTDs. Pregnant mice except controls were ip injected with LPS (25 µg/kg) daily from gestational day (GD)8 to GD12. In LPS+VitD3 group, pregnant mice were orally administered with VitD3 (25 µg/kg) before LPS injection. As expected, a 5-day LPS injection resulted in 62.5% (10/16) of dams and 20.3% of fetuses with NTDs. Additional experiment showed that a 5-day LPS injection downregulated placental proton-coupled folate transporter (pcft) and reduced folate carrier 1 (rfc1), 2 major folate transporters in placentas. Consistent with downregulation of placental folate transporters, folate transport from maternal circulation into embryos was disturbed in LPS-treated mice. Interestingly, VitD3 not only inhibited placental inflammation but also attenuated LPS-induced downregulation of placental folate transporters. Correspondingly, VitD3 markedly improved folate transport from maternal circulation into the embryos. Importantly, supplementation with VitD3 during pregnancy protected mice from LPS-induced NTDs. Taken together, these results suggest that supplementation with VitD3 during pregnancy prevents LPS-induced NTDs through inhibiting placental inflammation and improving folate transport from maternal circulation into the embryos.


Asunto(s)
Colecalciferol/administración & dosificación , Suplementos Dietéticos , Ácido Fólico/metabolismo , Lipopolisacáridos/toxicidad , Defectos del Tubo Neural/prevención & control , Placenta/metabolismo , Animales , Quimiocinas/metabolismo , Citocinas/metabolismo , Femenino , Intercambio Materno-Fetal , Ratones , Ratones Endogámicos ICR , Embarazo , Regulación hacia Arriba
9.
PLoS One ; 9(11): e113763, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25420102

RESUMEN

Lipopolysaccharide (LPS) has been associated with adverse pregnant outcomes, including fetal demise, intra-uterine growth restriction (IUGR), neural tube defects (NTDs) and preterm delivery in rodent animals. Previous studies demonstrated that melatonin protected against LPS-induced fetal demise, IUGR and preterm delivery. The aim of the present study was to investigate the effects of melatonin on LPS-induced NTDs. All pregnant mice except controls were intraperitoneally injected with LPS (25 µg/kg) daily from gestational day (GD)8 to GD12. Some pregnant mice were orally administered with melatonin (MT, 50 mg/kg) before each LPS injection. A five-day LPS injection resulted in 27.5% of fetuses with anencephaly, exencephaly or encephalomeningocele. Additional experiment showed that maternal LPS exposure significantly down-regulated placental proton-coupled folate transporter (pcft) and disturbed folate transport from maternal circulation through the placentas into the fetus. Interestingly, melatonin significantly attenuated LPS-induced down-regulation of placental pcft. Moreover, melatonin markedly improved the transport of folate from maternal circulation through the placentas into the fetus. Correspondingly, orally administered melatonin reduced the incidence of LPS-induced anencephaly, exencephaly or encephalomeningocele. Taken together, these results suggest that orally administered melatonin prevents LPS-induced NTDs through alleviating LPS-induced disturbance of folate transport from maternal circulation through the placenta into the fetus.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Melatonina/farmacología , Defectos del Tubo Neural/prevención & control , Placenta/metabolismo , Administración Oral , Anencefalia/inducido químicamente , Anencefalia/embriología , Anencefalia/prevención & control , Animales , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Quimiocinas/genética , Quimiocinas/metabolismo , Femenino , Ácido Fólico/sangre , Ácido Fólico/metabolismo , Mediadores de Inflamación/metabolismo , Lipopolisacáridos , Masculino , Intercambio Materno-Fetal/efectos de los fármacos , Melatonina/administración & dosificación , Meningocele/inducido químicamente , Meningocele/embriología , Meningocele/prevención & control , Ratones Endogámicos ICR , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/embriología , Embarazo , Transportador de Folato Acoplado a Protón/genética , Transportador de Folato Acoplado a Protón/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Toxicol Sci ; 138(2): 310-21, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24449419

RESUMEN

Increasing evidence demonstrates that cadmium (Cd) induces inflammation, but its mechanisms remain obscure. The present study showed that treatment with CdCl2 selectively upregulates macrophage inflammatory protein (MIP)-2 and cyclooxygenase (COX)-2 in RAW264.7 cells. Concomitantly, Cd²âº markedly elevated the level of phosphorylated Akt in dose- and time-dependent manners. LY294002, a specific inhibitor of phosphatidylinositol 3-kinase (PI3K), blocked Cd²âº-evoked Akt phosphorylation. Correspondingly, LY294002 significantly repressed Cd²âº-induced upregulation of MIP-2 and COX-2 in RAW264.7 cells. Further experiments showed that treatment with Cd²âº significantly reduced the level of PTEN protein in RAW264.7 cells. MG132, a specific proteasome inhibitor, blocked Cd²âº-induced reduction in PTEN protein as well as Akt phosphorylation, implicating the involvement of proteasome-mediated PTEN degradation. Of interest, Cd²âº-induced degradation of PTEN protein appears to be associated with PTEN ubiquitination. N-acetylcysteine, a glutathione (GSH) precursor, blocked Cd²âº-evoked PTEN degradation as well as Akt phosphorylation. By contrast, L-buthionine-S,R-sulfoximine, an inhibitor of cellular GSH synthesis, exacerbated Cd²âº-induced PTEN degradation and Akt phosphorylation. Alpha-phenyl-N-tert-butylnitrone and vitamin C, two antioxidants, did not prevent from Cd²âº-induced PTEN degradation and Akt phosphorylation. In conclusion, Cd²âº selectively induces MIP-2 and COX-2 through PTEN-mediated PI3K/Akt activation. Cellular GSH depletion mediates Cd²âº-induced PTEN degradation and subsequent PI3K/Akt activation in macrophages.


Asunto(s)
Cloruro de Cadmio/toxicidad , Quimiocina CXCL2/biosíntesis , Ciclooxigenasa 2/biosíntesis , Contaminantes Ambientales/toxicidad , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Técnicas de Cultivo de Célula , Línea Celular , Supervivencia Celular/efectos de los fármacos , Quimiocina CXCL2/genética , Ciclooxigenasa 2/genética , Relación Dosis-Respuesta a Droga , Inducción Enzimática , Glutatión/metabolismo , Immunoblotting , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Macrófagos/metabolismo , Ratones , Factores de Tiempo , Regulación hacia Arriba
11.
PLoS One ; 8(12): e82713, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24324824

RESUMEN

Increasing evidence demonstrates that maternal folic acid (FA) supplementation during pregnancy reduces the risk of neural tube defects, but whether FA prevents preterm delivery and intrauterine growth restriction (IUGR) remains obscure. Previous studies showed that maternal lipopolysaccharide (LPS) exposure induces preterm delivery, fetal death and IUGR in rodent animals. The aim of this study was to investigate the effects of FA on LPS-induced preterm delivery, fetal death and IUGR in mice. Some pregnant mice were orally administered with FA (0.6, 3 or 15 mg/kg) 1 h before LPS injection. As expected, a high dose of LPS (300 µg/kg, i.p.) on gestational day 15 (GD15) caused 100% of dams to deliver before GD18 and 89.3% of fetuses dead. A low dose of LPS (75 µg/kg, i.p.) daily from GD15 to GD17 resulted in IUGR. Interestingly, pretreatment with FA prevented LPS-induced preterm delivery and fetal death. In addition, FA significantly attenuated LPS-induced IUGR. Further experiments showed that FA inhibited LPS-induced activation of nuclear factor kappa B (NF-κB) in mouse placentas. Moreover, FA suppressed LPS-induced NF-κB activation in human trophoblast cell line JEG-3. Correspondingly, FA significantly attenuated LPS-induced upregulation of cyclooxygenase (COX)-2 in mouse placentas. In addition, FA significantly reduced the levels of interleukin (IL)-6 and keratinocyte-derived cytokine (KC) in amniotic fluid of LPS-treated mice. Collectively, maternal FA supplementation during pregnancy protects against LPS-induced preterm delivery, fetal death and IUGR through its anti-inflammatory effects.


Asunto(s)
Antiinflamatorios/farmacología , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/prevención & control , Ácido Fólico/farmacología , Lipopolisacáridos/efectos adversos , Nacimiento Prematuro/inducido químicamente , Nacimiento Prematuro/prevención & control , Sustancias Protectoras/farmacología , Animales , Antiinflamatorios/administración & dosificación , Línea Celular , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Suplementos Dietéticos , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Femenino , Muerte Fetal/inducido químicamente , Muerte Fetal/prevención & control , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/metabolismo , Ácido Fólico/administración & dosificación , Humanos , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/administración & dosificación , Masculino , Ratones , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Placenta/metabolismo , Embarazo , Nacimiento Prematuro/genética , Nacimiento Prematuro/metabolismo , Sustancias Protectoras/administración & dosificación , Trofoblastos/citología , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo
12.
J Pineal Res ; 53(4): 325-34, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22537289

RESUMEN

Increasing evidence demonstrates that melatonin has an anti-inflammatory effect. Nevertheless, the molecular mechanisms remain obscure. In this study, we investigated the effect of melatonin on toll-like receptor 4 (TLR4)-mediated molecule myeloid differentiation factor 88 (MyD88)-dependent and TRIF-dependent signaling pathways in lipopolysaccharide (LPS)-stimulated macrophages. RAW264.7 cells were incubated with LPS (2.0 µg/mL) in the absence or presence of melatonin (10, 100, 1000 µm). As expected, melatonin inhibited TLR4-mediated tumor necrosis factor alpha (TNF-α), interleukin (IL)-1ß, IL-6, IL-8, and IL-10 in LPS-stimulated macrophages. In addition, melatonin significantly attenuated LPS-induced upregulation of cyclooxygenase (COX)-2 and inducible nitric oxide synthase (iNOS) in macrophages. Further analysis showed that melatonin inhibited the expression of MyD88 in LPS-stimulated macrophages. Although it had no effect on TLR4-mediated phosphorylation of c-Jun N-terminal kinase (JNK), p38, and extracellular regulated protein kinase (ERK), melatonin significantly attenuated the activation of nuclear factor kappa B (NF-κB) in LPS-stimulated macrophages. In addition, melatonin inhibited TLR4-mediated Akt phosphorylation in LPS-stimulated macrophages. Moreover, melatonin significantly attenuated the elevation of interferon (IFN)-regulated factor-3 (IRF3), which was involved in TLR4-mediated TRIF-dependent signaling pathway, in LPS-stimulated macrophages. Correspondingly, melatonin significantly alleviated LPS-induced IFN-ß in macrophages. In conclusion, melatonin modulates TLR4-mediated inflammatory genes through MyD88-dependent and TRIF-dependent signaling pathways.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Antiinflamatorios/farmacología , Mediadores de Inflamación/metabolismo , Inflamación/prevención & control , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Melatonina/farmacología , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/efectos de los fármacos , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Línea Celular , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Activación Enzimática , Regulación de la Expresión Génica , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factor 88 de Diferenciación Mieloide/genética , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Factores de Tiempo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...